Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mod Pathol ; 28(9): 1174-84, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26321097

RESUMEN

Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive breast cancer and a frequent mammographic finding requiring treatment. Up to 25% of DCIS can recur and half of recurrences are invasive, but there are no reliable biomarkers for recurrence. We hypothesised that copy number aberrations could predict likelihood of recurrence. We analysed a cohort of pure DCIS cases treated only with wide local excision for genome-wide copy number and loss of heterozygosity using Affymetrix OncoScan MIP arrays. Cases included those without recurrence within 7 years (n = 25) and with recurrence between 1 and 5 years after diagnosis (n = 15). Pure DCIS were broadly similar in copy number changes compared with invasive breast cancer, with the consistent exception of a greater frequency of ERBB2 amplification in DCIS. There were no significant differences in age or ER status between the cases with a recurrence vs those without. Overall, the DCIS cases with recurrence had more copy number events than the DCIS without recurrence. The increased copy number appeared non-random with several genomic regions showing an increase in frequency in recurrent cases, including 20 q gain, ERBB2 amplification and 15q loss. Copy number changes may provide prognostic information for DCIS recurrence, but validation in additional cohorts is required.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma Intraductal no Infiltrante/genética , Recurrencia Local de Neoplasia/genética , Anciano , Femenino , Dosificación de Gen , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa
2.
PLoS Genet ; 8(9): e1002894, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23028338

RESUMEN

Despite intensive efforts using linkage and candidate gene approaches, the genetic etiology for the majority of families with a multi-generational breast cancer predisposition is unknown. In this study, we used whole-exome sequencing of thirty-three individuals from 15 breast cancer families to identify potential predisposing genes. Our analysis identified families with heterozygous, deleterious mutations in the DNA repair genes FANCC and BLM, which are responsible for the autosomal recessive disorders Fanconi Anemia and Bloom syndrome. In total, screening of all exons in these genes in 438 breast cancer families identified three with truncating mutations in FANCC and two with truncating mutations in BLM. Additional screening of FANCC mutation hotspot exons identified one pathogenic mutation among an additional 957 breast cancer families. Importantly, none of the deleterious mutations were identified among 464 healthy controls and are not reported in the 1,000 Genomes data. Given the rarity of Fanconi Anemia and Bloom syndrome disorders among Caucasian populations, the finding of multiple deleterious mutations in these critical DNA repair genes among high-risk breast cancer families is intriguing and suggestive of a predisposing role. Our data demonstrate the utility of intra-family exome-sequencing approaches to uncover cancer predisposition genes, but highlight the major challenge of definitively validating candidates where the incidence of sporadic disease is high, germline mutations are not fully penetrant, and individual predisposition genes may only account for a tiny proportion of breast cancer families.


Asunto(s)
Neoplasias de la Mama/genética , Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , RecQ Helicasas/genética , Eliminación de Secuencia/genética , Alelos , Exoma/genética , Exones , Proteína del Grupo de Complementación C de la Anemia de Fanconi/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Genoma Humano , Humanos , Linaje , Polimorfismo Genético , RecQ Helicasas/metabolismo , Análisis de Secuencia de ADN
3.
Hum Mutat ; 33(1): 95-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21990120

RESUMEN

There is strong evidence that overtly inactivating mutations in RAD51C predispose to hereditary breast and ovarian cancer but the prevalence of such mutations, and whether they are associated with a particular clinical phenotype, remains unclear. Resolving these questions has important implications for the implementation of RAD51C into routine clinical genetic testing. Consequently, we have performed a large RAD51C mutation screen of hereditary breast and ovarian cancer families, and the first study of unselected patients diagnosed with ovarian cancer. Our data confirm a consistent but low frequency (2/335 families) of inactivating RAD51C mutations among families with a history of both breast and ovarian cancer and an absence of mutations among breast cancer only families (0/1,053 families). Our data also provide support for the designation of the missense variant p.Gly264Ser as a moderate penetrance allele.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas de Unión al ADN/genética , Mutación Missense , Neoplasias Ováricas/genética , Adulto , Comorbilidad , Análisis Mutacional de ADN , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Mutación de Línea Germinal , Humanos , Persona de Mediana Edad , Linaje , Penetrancia , Factores de Riesgo
4.
Breast Cancer Res Treat ; 134(2): 543-7, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22580995

RESUMEN

KLLN is a p53 target gene with DNA binding function and represents a highly plausible candidate breast cancer predisposition gene. We screened for predisposing variants in 860 high-risk breast cancer families using high resolution melt analysis. A germline c.339_340delAG variant predicted to cause premature termination of the protein after 57 alternative amino acid residues was identified in 3/860 families who tested negative for BRCA1 and BRCA2 mutations and in 1/84 sporadic breast cancer cases. However, the variant was also detected in 2/182 families with known BRCA1 or BRCA2 mutations and in 2/464 non-cancer controls. Furthermore, loss of the mutant allele was detected in 2/2 breast tumors. Our data suggest that pathogenic mutations in KLLN are rare in breast cancer families and the c.339_340delAG variant does not represent a high-penetrance breast cancer risk allele.


Asunto(s)
Neoplasias de la Mama/genética , Predisposición Genética a la Enfermedad , Penetrancia , Proteínas Supresoras de Tumor/genética , Adulto , Secuencia de Bases , Estudios de Casos y Controles , Estudios de Cohortes , Análisis Mutacional de ADN , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Mutación de Línea Germinal , Humanos , Pérdida de Heterocigocidad , Datos de Secuencia Molecular , Eliminación de Secuencia
5.
BMC Cancer ; 11: 173, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21575258

RESUMEN

BACKGROUND: MAP2K4 is a putative tumor and metastasis suppressor gene frequently found to be deleted in various cancer types. We aimed to conduct a comprehensive analysis of this gene to assess its involvement in ovarian cancer. METHODS: We screened for mutations in MAP2K4 using High Resolution Melt analysis of 149 primary ovarian tumors and methylation at the promoter using Methylation-Specific Single-Stranded Conformation Polymorphism analysis of 39 tumors. We also considered the clinical impact of changes in MAP2K4 using publicly available expression and copy number array data. Finally, we used siRNA to measure the effect of reducing MAP2K4 expression in cell lines. RESULTS: In addition to 4 previously detected homozygous deletions, we identified a homozygous 16 bp truncating deletion and a heterozygous 4 bp deletion, each in one ovarian tumor. No promoter methylation was detected. The frequency of MAP2K4 homozygous inactivation was 5.6% overall, and 9.8% in high-grade serous cases. Hemizygous deletion of MAP2K4 was observed in 38% of samples. There were significant correlations of copy number and expression in three microarray data sets. There was a significant correlation between MAP2K4 expression and overall survival in one expression array data set, but this was not confirmed in an independent set. Treatment of JAM and HOSE6.3 cell lines with MAP2K4 siRNA showed some reduction in proliferation. CONCLUSIONS: MAP2K4 is targeted by genetic inactivation in ovarian cancer and restricted to high grade serous and endometrioid carcinomas in our cohort.


Asunto(s)
MAP Quinasa Quinasa 4/genética , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/genética , Proteínas Supresoras de Tumor/genética , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Bases , Proliferación Celular , Metilación de ADN/genética , Femenino , Dosificación de Gen/genética , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Genotipo , Humanos , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación/genética , Neoplasias Ováricas/mortalidad , Regiones Promotoras Genéticas/genética , Análisis de Supervivencia
6.
Breast Cancer Res Treat ; 119(2): 491-6, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19189213

RESUMEN

Heterozygous somatic mutations of the transcription factor, GATA-3, have recently been reported in approximately 5% breast of tumors unselected for family history. We sequenced the GATA-3 gene in 55 breast tumors from women with familial breast cancer, and found seven heterozygous somatic mutations, all in non-BRCA1/2 cases in which the frequency was 22%. In contrast, we found mutations of GATA-3 in only 4% of 81 sporadic tumors analysed. It is possible that GATA3 mutations occur earlier in the evolution of BRCAx tumors, compared to BRCA1, BRCA2 or sporadic tumors, and are therefore easier to detect by direct sequencing in the presence of some stromal contamination.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias de la Mama/genética , Factor de Transcripción GATA3/genética , Mutación , Proteínas Reguladoras de la Apoptosis , Neoplasias de la Mama/patología , Análisis Mutacional de ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Linaje , Medición de Riesgo , Factores de Riesgo
7.
Clin Cancer Res ; 14(22): 7246-50, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19010840

RESUMEN

PURPOSE: There is accumulating evidence that microRNAs may function like classic tumor suppressor genes but little is known about their mechanism of inactivation in cancer cells. We investigated whether somatic mutations are a common mechanism of inactivation of microRNA genes in ovarian cancer. EXPERIMENTAL DESIGN: Ten cancer-implicated microRNA genes were analyzed for somatic mutations in 90 ovarian epithelial cancers and matching normal DNA. High-resolution melt analysis and bidirectional sequencing was used to detect sequence variations. RESULTS: High-resolution melt analysis and direct sequencing did not identify any somatic mutations but did reveal numerous novel and previously reported germ line base substitutions, deletions, and insertions surrounding the mature microRNA sequences. The majority of variants were detected in the same proportion of non-cancer control individuals suggesting that they do not represent ovarian cancer-predisposing alleles. CONCLUSION: The absence of somatic mutations in any of the 10 cancer-implicated microRNAs in our large cohort of ovarian tumors suggests that this may be an uncommon mechanism of inactivation of microRNAs in ovarian cancer.


Asunto(s)
MicroARNs/genética , Neoplasias Ováricas/genética , ADN de Neoplasias/genética , Femenino , Humanos , Mutación , Reacción en Cadena de la Polimerasa
8.
Breast Cancer Res Treat ; 112(2): 351-2, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18095152

RESUMEN

Mutations in BRCA1 predispose to breast cancer. CTIP interacts with BRCA1 and so could also be associated with increased risk. We screened CTIP for germline mutations in 210 probands of breast cancer families including 129 families with no mutations in BRCA1 or BRCA2. No coding variants were detected in CTIP, therefore, it is unlikely to be involved in breast cancer risk.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA1/fisiología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Genes BRCA1 , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Australia , Neoplasias de la Mama/etnología , Análisis Mutacional de ADN , Endodesoxirribonucleasas , Salud de la Familia , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Tamizaje Masivo/métodos , Repeticiones de Microsatélite , Riesgo
9.
Clin Cancer Res ; 13(16): 4731-9, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17699850

RESUMEN

PURPOSE: Genetic changes in sporadic ovarian cancer are relatively poorly characterized compared with other tumor types. We have evaluated the use of high-resolution whole genome arrays for the genetic profiling of epithelial ovarian cancer. EXPERIMENTAL DESIGN: We have evaluated 31 primary ovarian cancers and matched normal DNA for loss of heterozygosity and copy number alterations using 500 K single nucleotide polymorphism arrays. RESULTS: In addition to identifying the expected large-scale genomic copy number changes, >380 small regions of copy number gain or loss (<500 kb) were identified among the 31 tumors, including 33 regions of high-level gain (>5 copies) and 27 homozygous deletions. The existence of such a high frequency of small regions exhibiting copy number alterations had not been previously suspected because earlier genomic array platforms lacked comparable resolution. Interestingly, many of these regions harbor known cancer genes. For example, one tumor harbored a 350-kb high-level amplification centered on FGFR1 and three tumors showed regions of homozygous loss 109 to 216 kb in size involving the RB1 tumor suppressor gene only. CONCLUSIONS: These data suggest that novel cancer genes may be located within the other identified small regions of copy number alteration. Analysis of the number of copy number breakpoints and the distribution of the small regions of copy number change indicate high levels of structural chromosomal genetic instability in ovarian cancer.


Asunto(s)
Aberraciones Cromosómicas , Dosificación de Gen , Neoplasias Glandulares y Epiteliales/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/genética , Polimorfismo de Nucleótido Simple , Desequilibrio Alélico , Cromosomas Humanos Par 9 , Femenino , Amplificación de Genes , Eliminación de Gen , Humanos , MAP Quinasa Quinasa 4/genética
10.
Cancer Lett ; 247(1): 91-7, 2007 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-16644110

RESUMEN

Chromosome 22q shows a high frequency of loss of heterozygosity (LOH) in ovarian cancers suggesting the existence of one or more important tumor suppressor genes (TSGs). The tissue inhibitor of metalloproteinase-3 (TIMP-3) is a plausible TSG candidate since it is often encompassed within these regions of LOH. TIMP-3 has not previously been investigated for somatic mutations or promoter hypermethylation in ovarian cancer. We analyzed 65 ovarian cancers for both somatic genetic mutations and TIMP-3 promoter hypermethylation. Screening of all coding exons of TIMP-3 did not reveal any somatic genetic mutations and only 1/65 showed TIMP-3 methylation. Our data indicate that inactivation of TIMP-3 by somatic mutation or promoter hypermethylation is rare in ovarian cancer.


Asunto(s)
Metilación de ADN , Genes Supresores de Tumor , Pérdida de Heterocigocidad , Mutación , Neoplasias Ováricas/genética , Inhibidores Tisulares de Metaloproteinasas/genética , Secuencia de Bases , Cromosomas Humanos Par 22 , Femenino , Humanos , Datos de Secuencia Molecular , Polimorfismo Conformacional Retorcido-Simple , Regiones Promotoras Genéticas , Inhibidor Tisular de Metaloproteinasa-4
11.
Clin Cancer Res ; 12(12): 3713-5, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16778097

RESUMEN

PURPOSE: A very high frequency of somatic mutations in the transforming growth factor-beta signaling component km23 has been reported in a small series of ovarian cancers (8 of 19, 42%). Functional studies showed that some mutations disrupt km23 function, resulting in aberrant transforming growth factor-beta signaling and presumably enhanced tumorigenicity. If verified, this would elevate mutation of km23 as the single most frequent somatic event in ovarian cancer. EXPERIMENTAL DESIGN: We sought to verify the frequency of silencing of km23 among 104 primary ovarian cancers (49 serous, 18 mucinous, 29 endometrioid/clear cell, and 8 undifferentiated) as well as 72 breast and 61 colorectal cancers by undertaking both somatic mutation and promoter methylation analyses. All four exons of km23 were individually amplified from genomic DNA with primers complementary to surrounding intronic sequences and analyzed by single-stranded conformational polymorphism analysis. RESULTS: Two germ line polymorphisms were identified, but none of the 237 tumors analyzed harbored somatic km23 mutations. In addition, promoter methylation analysis showed that in all cases, the 5' CpG island was unmethylated. CONCLUSIONS: Our data suggest that silencing of km23, either through somatic genetic mutation or promoter hypermethylation, is rare in ovarian, breast, and colorectal cancers.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Colorrectales/genética , Dineínas/genética , Neoplasias Ováricas/genética , Secuencia de Bases , Neoplasias de la Mama/prevención & control , Neoplasias Colorrectales/prevención & control , Dineínas Citoplasmáticas , Cartilla de ADN , Femenino , Silenciador del Gen , Humanos , Mutación , Neoplasias Ováricas/prevención & control , Regiones Promotoras Genéticas , Proteínas Supresoras de Tumor/genética
12.
Cancer Lett ; 240(2): 195-7, 2006 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-16239061

RESUMEN

A functional T to G germline polymorphism in the promoter region of MDM2 (SNP309) has been reported to profoundly accelerate tumor formation suggesting that it may also represent a powerful cancer predisposing allele. To investigate the role of SNP309 in cancer predisposition we undertook a case-control study of this polymorphism among 351 women diagnosed with breast cancer, 302 women diagnosed with ovarian and 258 female controls from a British population. The GG genotype was not associated with either breast cancer (OR 1.04, 95% CI 0.67-1.60) or ovarian cancer (OR 0.86, 95% CI 0.53-1.37). This study has found no evidence that the GG genotype of the MDM2 SNP309 polymorphism is associated with early onset or familial breast cancer or with ovarian cancer.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias Ováricas/genética , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-mdm2/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Femenino , Humanos , Persona de Mediana Edad , Factores de Riesgo
13.
Cancer Lett ; 233(1): 151-7, 2006 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-15876481

RESUMEN

LZTS1 has been shown to have tumour suppressor activities against prostate and breast cancer and is located within a region of frequent loss of heterozygosity (LOH) at 8p22 in ovarian cancer. We have analysed the expression of LZTS1 in ovarian cancer and found no evidence of loss of expression relative to normal ovarian surface epithelial cells. We have also analysed the coding region of the LZTS1 gene in 87 primary ovarian adenocarcinomas by DHPLC and detected a single silent somatic mutation. These data indicate that LZTS1 is not the target of LOH at 8p22 in ovarian cancer.


Asunto(s)
Adenocarcinoma/genética , Proteínas de Unión al ADN/genética , Mutación , Neoplasias Ováricas/genética , Proteínas Supresoras de Tumor/genética , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Femenino , Humanos , Pérdida de Heterocigocidad , Ovario/metabolismo , Polimorfismo de Nucleótido Simple
14.
Cancer Res ; 64(21): 7678-81, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15520168

RESUMEN

Phosphatidylinositol 3'-kinases are lipid kinases with important roles in neoplasia. Recently, a very high frequency of somatic mutations in PIK3CA has been reported among a large series of colorectal cancers. However, the relevance of PIK3CA mutation in other cancer types remains unclear because of the limited number of tumors investigated. We have screened a total of 284 primary human tumors for mutations in all coding exons of PIK3CA using a combination of single stranded conformational polymorphism and denaturing high-performance liquid chromatography analysis. Among 70 primary breast cancers, 40% (28 of 70) harbored mutations in PIK3CA, making it the most common mutation described to date in this cancer type. Mutations were not associated with histologic subtype, estrogen receptor status, grade or presence of tumor in lymph nodes. Among the primary epithelial ovarian cancers only 11 of 167 (6.6%) contain somatic mutations, but there was a clear histologic subtype bias in their distribution. Only 2 of 88 (2.3%) of serous carcinomas had PIK3CA mutations compared with 8 of 40 (20.0%) endometrioid and clear cell cancers, which was highly significant (P = 0.001). In contrast, PIK3CA gene amplification (>7-fold) was common among all histologic subtypes (24.5%) and was inversely associated with the presence of mutations. Overall, PIK3CA mutation or gene amplification was detected in 30.5% of all ovarian cancers and 45% of the endometrioid and clear cell subtypes. Our study is the first direct evidence that PIK3CA is an oncogene in ovarian cancer and greatly extends recent findings in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Mutación , Neoplasias Ováricas/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasa Clase I , Exones , Femenino , Amplificación de Genes , Humanos , Fosfatidilinositol 3-Quinasas/fisiología
15.
Oncogene ; 23(46): 7726-33, 2004 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-15361843

RESUMEN

Suppressor of cytokine signaling (SOCS) proteins have emerged as critical attenuators of cytokine-mediated processes, suggesting a role in the suppression of tumorigenesis. In the ovary and mammary gland, cytokines such as prolactin and IL-6 are important regulators of growth and differentiation. We have investigated whether silencing or inactivation of SOCS genes occurs in ovarian and breast carcinomas. The SOCS1 and SOCS2 CpG islands were found to be hypermethylated in 23 and 14% of primary ovarian cancers, respectively, whereas only SOCS1 was methylated in breast cancers (9%). Methylation of these genes did not occur in normal tissues. No correlation was apparent between methylation and loss of heterozygosity, and no somatic mutations were found in a large panel of carcinomas. Aberrant methylation of these SOCS genes correlated with transcriptional silencing in ovarian and breast cancer cell lines, since expression was induced by the demethylating agent 5-azadeoxycytidine. SOCS3 was not hypermethylated in either cancer type. Consistent with this data, SOCS1 and SOCS2 but not SOCS3 suppressed the growth of ovarian and breast cancer cells. Hypermethylation and silencing of specific SOCS genes in the ovary, and to a lesser extent in breast, may augment cytokine responsiveness in these tissues, thereby contributing to oncogenesis.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Portadoras/genética , Metilación de ADN , Proteínas de Unión al ADN/genética , Péptidos y Proteínas de Señalización Intracelular , Neoplasias Ováricas/genética , Proteínas Represoras/genética , Transactivadores/genética , Factores de Transcripción/genética , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Femenino , Humanos , Pérdida de Heterocigocidad , Proteína 1 Supresora de la Señalización de Citocinas , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas
16.
Oncogene ; 22(32): 5070-81, 2003 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-12902990

RESUMEN

EDD (E3 isolated by differential display), located at chromosome 8q22.3, is the human orthologue of the Drosophila melanogaster tumour suppressor gene 'hyperplastic discs' and encodes a HECT domain E3 ubiquitin protein-ligase. To investigate the possible involvement of EDD in human cancer, several cancers from diverse tissue sites were analysed for allelic gain or loss (allelic imbalance, AI) at the EDD locus using an EDD-specific microsatellite, CEDD, and other polymorphic microsatellites mapped in the vicinity of the 8q22.3 locus. Of 143 cancers studied, 38 had AI at CEDD (42% of 90 informative cases). In 14 of these cases, discrete regions of imbalance encompassing 8q22.3 were present, while the remainder had more extensive 8q aberrations. AI of CEDD was most frequent in ovarian cancer (22/47 informative cases, 47%), particularly in the serous subtype (16/22, 73%), but was rare in benign and borderline ovarian tumours. AI was also common in breast cancer (31%), hepatocellular carcinoma (46%), squamous cell carcinoma of the tongue (50%) and metastatic melanoma (18%). AI is likely to represent amplification of the EDD gene locus rather than loss of heterozygosity, as quantitative RT-PCR and immunohistochemistry showed that EDD mRNA and protein are frequently overexpressed in breast and ovarian cancers, while among breast cancer cell lines EDD overexpression and increased gene copy number were correlated. These results demonstrate that AI at the EDD locus is common in a diversity of carcinomas and that the EDD gene is frequently overexpressed in breast and ovarian cancer, implying a potential role in cancer progression.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Ováricas/genética , Péptido Sintasas/genética , Ubiquitina-Proteína Ligasas , Aberraciones Cromosómicas , Cromosomas Humanos Par 8 , Femenino , Humanos , Repeticiones de Microsatélite , Neoplasias/genética , Péptido Sintasas/biosíntesis
17.
BMC Med Genomics ; 8: 45, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26231170

RESUMEN

BACKGROUND: Loss of heterozygosity (LOH) is a common genetic event in cancer development, and is known to be involved in the somatic loss of wild-type alleles in many inherited cancer syndromes. The wider involvement of LOH in cancer is assumed to relate to unmasking a somatically mutated tumour suppressor gene through loss of the wild type allele. METHODS: We analysed 86 ovarian carcinomas for mutations in 980 genes selected on the basis of their location in common regions of LOH. RESULTS: We identified 36 significantly mutated genes, but these could only partly account for the quanta of LOH in the samples. Using our own and TCGA data we then evaluated five possible models to explain the selection for non-random accumulation of LOH in ovarian cancer genomes: 1. Classic two-hit hypothesis: high frequency biallelic genetic inactivation of tumour suppressor genes. 2. Epigenetic two-hit hypothesis: biallelic inactivation through methylation and LOH. 3. Multiple alternate-gene biallelic inactivation: low frequency gene disruption. 4. Haplo-insufficiency: Single copy gene disruption. 5. Modified two-hit hypothesis: reduction to homozygosity of low penetrance germline predisposition alleles. We determined that while high-frequency biallelic gene inactivation under model 1 is rare, regions of LOH (particularly copy-number neutral LOH) are enriched for deleterious mutations and increased promoter methylation, while copy-number loss LOH regions are likely to contain under-expressed genes suggestive of haploinsufficiency. Reduction to homozygosity of cancer predisposition SNPs may also play a minor role. CONCLUSION: It is likely that selection for regions of LOH depends on its effect on multiple genes. Selection for copy number neutral LOH may better fit the classic two-hit model whereas selection for copy number loss may be attributed to its effect on multi-gene haploinsufficiency. LOH mapping alone is unlikely to be successful in identifying novel tumour suppressor genes; a combined approach may be more effective.


Asunto(s)
Pérdida de Heterocigocidad , Neoplasias Ováricas/genética , Femenino , Dosificación de Gen/genética , Genes Relacionados con las Neoplasias/genética , Genes Supresores de Tumor , Genómica , Haploinsuficiencia/genética , Humanos
18.
PLoS One ; 7(4): e35805, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22536442

RESUMEN

MicroRNAs are key regulators of gene expression and have been shown to have altered expression in a variety of cancer types, including epithelial ovarian cancer. MiRNA function is most often achieved through binding to the 3'-untranslated region of the target protein coding gene. Mutation screening using massively-parallel sequencing of 712 miRNA genes in 86 ovarian cancer cases identified only 5 mutated miRNA genes, each in a different case. One mutation was located in the mature miRNA, and three mutations were predicted to alter the secondary structure of the miRNA transcript. Screening of the 3'-untranslated region of 18 candidate cancer genes identified one mutation in each of AKT2, EGFR, ERRB2 and CTNNB1. The functional effect of these mutations is unclear, as expression data available for AKT2 and EGFR showed no increase in gene transcript. Mutations in miRNA genes and 3'-untranslated regions are thus uncommon in ovarian cancer.


Asunto(s)
Regiones no Traducidas 3' , MicroARNs/genética , Mutación , Neoplasias Ováricas/genética , Sitios de Unión/genética , Análisis Mutacional de ADN , Receptores ErbB/genética , Femenino , Estudios de Asociación Genética , Humanos , Proteínas Proto-Oncogénicas c-akt/genética , Receptor ErbB-2/genética , beta Catenina/genética
20.
Breast Cancer Res Treat ; 111(3): 505-9, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17972171

RESUMEN

Several studies in various populations have suggested that non-synonymous BARD1 variants are associated with increased breast cancer risk. Using DHPLC analysis we screened the coding region of BARD1 for variants in 210 probands of breast cancer families including 129 families with no mutations in BRCA1 or BRCA2. These families were ascertained in Australia through the Kathleen Cunningham Foundation Consortium for Research into Familial Breast Cancer (kConFab). Nine coding variants were detected among the kConFab families, including two novel variants (Thr598Ile and Ile692Thr). The frequency of five of these variants were evaluated in 258 non-cancer controls and 401 women with sporadic breast cancer. Three variants (1139del21, G1756C and A2285G) were detected in all three groups at a similar frequency suggesting that these do not represent BRCAX candidates. Two variants (Thr598Ile and Ile692Thr) were not detected in any of the 659 sporadic breast cancer cases and controls and were assessed for segregation with breast cancer in the families of the probands. However, neither variant was identified in any other breast cancer case in either family suggesting that these variants are non-pathogenic polymorphisms. We have found no evidence to support involvement of BARD1 in familial breast cancer risk in the Australian population. In addition, three variants previously reported to be pathogenic in other populations are likely to represent benign polymorphisms and therefore we conclude that BARD1 is unlikely to represent a high-penetrance breast cancer susceptibility gene.


Asunto(s)
Neoplasias de la Mama/genética , Polimorfismo Genético , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Australia , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Linaje , Medición de Riesgo , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA